EP3263567A1 - Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités - Google Patents

Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités Download PDF

Info

Publication number
EP3263567A1
EP3263567A1 EP16177663.8A EP16177663A EP3263567A1 EP 3263567 A1 EP3263567 A1 EP 3263567A1 EP 16177663 A EP16177663 A EP 16177663A EP 3263567 A1 EP3263567 A1 EP 3263567A1
Authority
EP
European Patent Office
Prior art keywords
compound
stands
phenyl
group
halogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP16177663.8A
Other languages
German (de)
English (en)
Inventor
designation of the inventor has not yet been filed The
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aicuris Anti Infectives Cures GmbH
Original Assignee
Aicuris Anti Infectives Cures GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aicuris Anti Infectives Cures GmbH filed Critical Aicuris Anti Infectives Cures GmbH
Priority to EP16177663.8A priority Critical patent/EP3263567A1/fr
Publication of EP3263567A1 publication Critical patent/EP3263567A1/fr
Ceased legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to carboxamide-substituted pyrazoles and tri(hetero)aryl pyrazoles, pharmaceutical formulations containing such compounds, processes for manufacture and the use of such compounds in methods of reducing the heart rate and, accordingly, for its use in methods of treating and/or preventing cardiovascular diseases and/or cardiovascular comorbidities, which are exacerbated by an increased average 24-h heart rate of ⁇ 90 beats/min.
  • bpm cardiovascular diseases selected from the group comprising cardiac arrhythmia (hereinafter CA), hypertensive heart disease (hereinafter HHD), tachycardia, inappropriate sinus tachycardia (hereinafter IST), angina pectoris (hereinafter AP) and stable angina pectoris (hereinafter SAP), myocardial ischemia (hereinafter MI), coronary artery disease (hereinafter CAD), heart failure (hereinafter HF), and stroke, as well as cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and chronic kidney disease (hereinafter CKD) in humans and/or animals.
  • CA cardiac arrhythmia
  • HHD hypertensive heart disease
  • IST inappropriate sinus tachycardia
  • AP angina pectoris
  • SAP stable angina pectoris
  • MI myocardial ischemia
  • CAD coronary artery disease
  • HF heart failure
  • stroke as well as cardiovascular comorbidities selected from the group comprising diabetes, metabolic
  • CVD Cardiovascular diseases
  • CVDs are a class of diseases that involve the heart and/or blood vessels.
  • CVDs generally include coronary artery diseases (CAD(s)) such as AP and myocardial infarction (also known as heart attack).
  • CAD(s) coronary artery diseases
  • Other CVDs are inter alia CA, HHD, IST, AP and SAP, MI, HF, and stroke.
  • CVDs may vary depending on the disease concerned. For instance, CAD and stroke involve atherosclerosis. This may be caused by high blood pressure, smoking, diabetes, lack of exercise, obesity, high blood cholesterol, poor diet, and excessive alcohol consumption, among others. Of note, high blood pressure results in 13 % of CVD deaths.
  • CVDs involving the blood vessels are known as vascular diseases and are reflected by inter alia CAD, peripheral arterial disease (disease of blood vessels that supply blood to the arms and legs), cerebrovascular disease (disease of blood vessels that supply blood to the brain, including stroke), renal artery stenosis, and aortic aneurysm.
  • CVDs that involve the heart are reflected by cardiomyopathy (diseases of cardiac muscle), HHD (diseases of the heart secondary to high blood pressure or hypertension), HF, pulmonary heart disease (a failure at the right side of the heart with respiratory system involvement), cardiac dysrhythmias (abnormalities of heart rhythm).
  • cardiomyopathy diseases of cardiac muscle
  • HHD diseases of the heart secondary to high blood pressure or hypertension
  • HF pulmonary heart disease
  • cardiac dysrhythmias abnormalities of heart rhythm
  • Dyslipidemia is associated with more than half of all cases of ischemic heart disease globally. About 17 million deaths occur worldwide each year from CVD. About 26 million people worldwide suffer from HF, accounting for 1 % - 2 % of healthcare resources in industrialized countries.
  • full treatment goals are not met with current therapeutics, such as beta blockers, angiotensinconverting-enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs), and diuretics in all HF patients.
  • Heart rate is an important contributor in the pathophysiology of CVDs, as e.g. in case of CAD and HF, and is being increasingly recognised as a modifiable risk factor in respective patients.
  • Heart rate is determined by spontaneous electrical pacemaker activity in the sinoatrial node (hereinafter SAN). Cardiac pacemaker cells generate the spontaneous slow diastolic depolarisation that drives the membrane voltage away from a hyperpolarised level towards the threshold level for initiating a subsequent action potential, generating rhythmic action potentials that propagate through the heart and trigger myocardial contraction.
  • the rate of spontaneous diastolic depolarization is significantly influenced by l f , a mixed sodium-potassium current involving ion movement across the so-called f-channels, which are activated by hyperpolarization, and the opening of which is dependent on the intracellular availability of cyclic adenosine monophosphate.
  • the l f current is thus an ionic current that determines the slope of the diastolic depolarisation, which in turn controls the heart rate.
  • the cardiac impulse originates in the SAN, located at the right atrial endocardium, between the upper and lower cava vein, and formed by highly specialized cells that are able to generate action potentials, which start the sinus rhythm.
  • phase 4 is characterized by a slow increase in membrane potential to the threshold for triggering a new action potential.
  • currents both inward and outward involved in the functioning of the SAN, able to determine the profile of the final current.
  • the three main ones are:
  • the funny current i.e. l f
  • the funny current is highly expressed in spontaneously active cardiac regions, such as the SAN, which is the natural pacemaker region, the atrioventricular node and the Purkinje fibres of conduction tissue.
  • the funny current is a mixed sodium-potassium current that activates upon hyperpolarization at voltages in the diastolic range (normally from - 60/- 70 mV to - 40 mV).
  • the membrane When at the end of a sinoatrial action potential the membrane repolarizes below the l f threshold (about - 40/ - 50 mV), the funny current is activated and supplies inward current, which is responsible for starting the diastolic depolarization phase.
  • the funny current controls the rate of spontaneous activity of sinoatrial myocytes, and hence the cardiac rate.
  • HCN hyperpolarization-activated cyclic nucleotide-gated channels family
  • Kv voltage-gated K +
  • CNG CNG channels
  • HCN channels are a family of six transmembrane domain, single pore-loop, hyperpolarization activated, non-selective cation channels.
  • HCN4 is considered the main isoform that is able to control the heart rate.
  • HCN4 channels may be coupled to rhythm disturbances, which have been already reported in humans.
  • an inherited mutation of a highly conserved residue in the CNBD of the HCN4 protein (S672R) is associated with inherited sinus bradycardia.
  • a missense mutation (S672R) on the exon 7 in hHCN4 gene induces the production of an isoform of the pacemaker channel changed at cAMP binding site level, which was responsible for a familial form of an asymptomatic bradycardia.
  • l f generates spontaneous activity and modulates cardiac rate.
  • activation is the key process in the modulation of rate by autonomic transmitters.
  • the mammalian pacemaker region (SAN) is densely innervated by the autonomic nervous system: sympathetic ß-adrenergic stimulation accelerates, and parasympathetic muscarinic stimulation slows cardiac rate.
  • l f inhibition may lead to slowing down cardiac rate.
  • pharmacological reduction of heart rate is a crucial target in the treatment of several pathological conditions characterized by a deficiency of oxygen supply to the working myocardium, such as AP, a common manifestation of CAD, MI and HF.
  • HTN4 heart rate-inhibiting agents
  • cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke
  • cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD in humans and/or animals.
  • ivabradine a new molecule selected for higher funny channel selectivity and reduced side effects, has been developed and is now being marketed for pharmacological treatment of chronic stable angina. Ivabradine is currently already used as a treatment option for SAP and HF in patients.
  • Ivabradine is a pure heart rate lowering agent acting by inhibiting the funny current (l f ) in the SAN.
  • Ivabradine is thus selective for the l f current and exerts significant inhibition thereof and heart rate reduction at concentrations that do not affect other cardiac ionic currents. Ivabradine lowers heart rate without any negative inotropic or lusitropic effect, thus preserving ventricular contractility. Ivabradine does not affect blood pressure, myocardial contractility, intra-cardiac conduction, or ventricular repolarisation.
  • Efficacy of ivabradine in four patients with inappropriate sinus tachycardia a three month-long experience based on electrocardiographic, Holter monitoring, exercise tolerance and quality of life assessments.
  • Kaplinsky E1 Comes FP, Urondo LS, Ayma FP; Heart Rhythm. 2010 Sep;7(9):1318-23 .
  • ivabradine is generally well tolerated, patients may still complain of side effects, such as visual symptoms and bradycardia.
  • ivabradine is contraindicated in severe bradycardia, II or III degree atrio-ventricular block, ejection fraction ⁇ 35%, and severe hypotension.
  • CYP 3A4 inhibitors such as ketoconazole, macrolides (such as erythromycin), and nefazodone (used in the treatment of human immunodeficiency virus).
  • beta blocking medication or calcium antagonists are still routinely administered prior to computer tomography coronary angiography (CTCA) examinations.
  • CTCA computer tomography coronary angiography
  • Ivabradine appears to be an alternative, but still provides for the above-mentioned side effects.
  • the use of beta blockers, calcium antagonists, or ivabradine is limited, however, due to various contraindications. In recent studies, for instance, a substantial portion of patients requiring heart rate reduction has contraindications to beta blockers.
  • the herein disclosed compounds of the classes carboxamide-substituted pyrazoles and tri(hetero)arylpyrazoles according to the general formula (I) are very potent and effective HCN4 blockers/inhibitors, and are thus useful in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, as well as to cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD in humans and/or animals.
  • the present invention is also directed to compounds of the general formula (I) for use in a method of treatment and/or prophylaxis of metabolic syndrome, diabetic complications, atherosclerosis, vascular dimensia, coronary heart disease, or congestive heart failure in a human and/or animal by administering to a human and/or animal in need of such treatment a therapeutically effective amount of a compound of the general formula (I) or a pharmaceutically acceptable salt of said compound.
  • the herein disclosed compounds of the general formula (I) are intended for the use in the aforementioned cardiovascular diseases and / or comorbidities thereof.
  • the inventors found that the compounds of the general formula (I) unexpectedly do not interact with other ion channels of Nav, Kav, and Cav substantially (as exemplarily shown for 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]-5,6- dihydro-7H-pyrrolo[3,4-d] pyrimidin-7-one, i.e. compound A in accordance with the invention; see further below).
  • the herein disclosed carboxamide-substituted pyrazoles and tri(hetero)arylpyrazoles may be effective in lowering the heart rate in humans and animals, and may thus be exploited for its use in methods of treating and/or preventing the aforementioned cardiovascular diseases and / or comorbidities thereof.
  • the substance classes of carboxamide-substituted pyrazoles and tri(hetero)aryl-pyrazoles differ greatly from known HCN inhibitors (i.e. ivabradine, alinidine, zatebradine, cilobradine), and thus the compounds of general formula (I) in accordance with the invention may react in the methods of treatment and/or prevention as recited herein with different binding pockets and mechanisms than the compounds known in the art.
  • Tri(hetero)aryl-pyrazoles and analogues thereof as well as derivatives thereof are known in the art, and are described inter alia in DE 10 2012 016 908 ; WO 2014/027112 ; WO 2015/121413 ; WO 2013/079586 ; DE 10 2011 055 815 ; DE 10 2009 036 604 ; WO 2011/012630 ; DE 10 2008 015 032 ; DE 10 2008 015 033 ; WO 2009/115213 ; WO 2010/075962 ; DE 10 2008 062863 ; DE 10 2008 062 878 ; WO 2010/075957 ; DE 10 2009 038 123 ; and WO 2011/020822 .
  • WO 2011/058149 describes tricyclic pyrazole derivatives as PI3k inhibitors for the treatment of autoimmune diseases.
  • WO 2008/074982 describes pyrazole derivatives as CB1 receptor modulators in the treatment of overweight.
  • Pyrazole derivatives as agents against blood platelet aggregation for the treatment of ischemic diseases are described in WO 2004/069824 and WO 2006/004027 .
  • Pyrazole derivatives as COX-1 inhibitors are described in WO 2004/050632 and US 2004/0116475 .
  • WO 2008/017932 describes various aryl sulfonamides, among them a pyrazole-containing example, as carbonic anhydrase inhibitors.
  • DE 10 2008 015 033 and DE 10 2008 015 032 describe phenyl-substituted pyrazoles and their use for the treatment and prevention of infections with retroviruses.
  • subject matter of the present invention are the compounds of the represented formula (I), wherein said compound is i.e. 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-1-one, and their co-crystals or the polymorphs of their co-crystals, and their salts, their solvates or the solvates of their salts for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome
  • subject matter of the invention are the compounds of the represented formula (I), wherein said compound is i.e. 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]-5,6-dihydro-2-methyl-7H-pyrrolo[3,4-d] pyrimidin-7-one, and their co-crystals or the polymorphs of their co-crystals, and their salts, their solvates or the solvates of their salts for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes
  • subject matter of the invention are the compounds of the represented formula (I), wherein said compound is i.e. 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(3-cyano-4-fluoro-phenyl)-1 H-pyrazol-3-yl]-5,6-dihydro-7H-pyrrolo[3,4-d] pyrimidin-7-one, and their co-crystals or the polymorphs of their co-crystals, and their salts, their solvates or the solvates of their salts for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group
  • subject matter of the invention are the compounds of the represented formula (I), wherein said compound is i.e. 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]-5,6-dihydro-7H-pyrrolo[3,4-d]pyrimidin-7-one, and their co-crystals or the polymorphs of their co-crystals, and their salts, their solvates or the solvates of their salts for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome,
  • subject matter of the invention are the compounds of the represented formula (I), wherein said compound is i.e. 1-[[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]carbonyl]-imidazolidin-4-one, and their co-crystals or the polymorphs of their co-crystals, and their salts, their solvates or the solvates of their salts for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD.
  • cardiovascular diseases selected from the group comprising CA, H
  • subject matter of the invention are the compounds of the represented formula (I), wherein said compound is i.e. 3-Amino-4-[5-[3-chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl-carbonyl]-1,5-dihydro-2H-pyrrol-2-one, and their co-crystals or the polymorphs of their co-crystals, and their salts, their solvates or the solvates of their salts for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity
  • the present invention comprises any tautomeric form for its use in a method of treating and/preventing the aforementioned cardiovascular diseases and / or cardiovascular comorbidities thereof.
  • Isomeres are to be understood as tautomeres or tautomeric forms within the scope of the present invention, which are characterized in that said isomeres have the same chemical formula as their original compound in accordance with the respective formula (I) or their co-crystals or the polymorphs of their co-crystals, or their salts, their solvates or the solvates of their salts; isolated atoms, however, can be positionally arranged differently.
  • these isomers are therefore described as tautomeres or tautomeric forms because they can quickly interconvert into each other through the migration of isolated atoms or atom groups, and, in so doing, the respective isomers can be in a rapidly changing chemical equilibrium with one another.
  • the tautomers or tautomeric forms thus only differ in the position of a chemical group and in the position of a double bond and can be understood as the same chemical compound in accordance with the formula (I) or their co-crystals or the polymorphs of their co-crystals, or their salts, their solvates or the solvates of their salts.
  • salts within the scope of the present invention, physiologically acceptable salts of the compounds pursuant to the invention are preferred.
  • salts which are not suitable for pharmaceutical administration itself, but can, for example, be used for the isolation or cleaning of the compounds in accordance with the invention.
  • Physiologically acceptable salts of the compounds of the invention include acid addition salts of mineral acids, carboxylic acids, and sulfonic acids, e.g. salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methane sulfonic acid, ethane sulfonic acid, p-toluene sulfonic acid, benzene sulfonic acid, naphthalene disulfonic acid, acetic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
  • sulfonic acids e.g. salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methane sulfonic acid, ethane sulfonic acid, p-toluene sulfonic acid, benzene sulfonic acid,
  • Physiologically acceptable salts of the compounds in accordance with the inventions also comprise salts of customary bases, such as exemplary and preferably alkali metal salts (e.g., sodium and potassium salts), alkaline earth metal salts (e.g., calcium and magnesium salts) and ammonium salts, derived from ammonia or organic amines with 1 to 16 C atoms, such as exemplarily and preferably ethylamine, diethyl amine, triethyl amine, ethyl diisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N -methylmorpholine, arginine, lysine, ethylendiamine and N -methylpiperidine.
  • customary bases such as exemplary and preferably alkali metal salts (e.g., sodium and potassium salts), alkaline earth metal salts (e.g
  • Solvates within the scope of the invention are those forms of the compounds in accordance with the invention that in a solid or liquid state forms a complex through coordination with solvent molecules. Hydrates are a special form of the solvate in which the coordination takes place with water.
  • Co-crystals within the scope of the invention are those solid forms of the compounds in accordance with the invention that are crystalline single phase materials composed of two or more different molecular and/or ionic compounds generally in a stoichiometric ratio which are neither solvates nor simple salts as defined above.
  • the difference between a crystalline salt and a co-crystal lies in the transfer of a proton.
  • the transfer of protons from one component to another in a crystal is dependent on the environment.
  • crystalline salts and co-crystals may be thought of in accordance with the invention as two ends of a proton transfer spectrum, where the salt has completed the proton transfer at one end and an absence of proton transfer exists for co-crystals at the other end.
  • Polymorphs of co-crystals within the scope of the invention are those solid forms of the above-defined co-crystals, which, however, exist in more than one form or co-crystal structure. Said polymorphs of co-crystals may vary in certain morphologies of those solid forms, such as the variables of crystal habit, amorphous fraction or crystallographic defects.
  • Alkoxy within the scope of the invention preferably stands for a branched or unbranched alkoxy residue, in particular with 1 to 6, 1 to 4 or 1 to 3 carbon atoms.
  • Preferred is an unbranched or branched alkoxy residue with 1 to 3 carbon atoms.
  • reference is made to: methoxy, ethoxy, n-propoxy, isopropoxy, t-butoxy, n-pentoxy and n-hexoxy.
  • Halogen stands for fluorine, chlorine, bromine or iodine, with fluorine and chlorine being preferred, unless otherwise specified.
  • the conversion generally takes place in two stages in inert solvents, first forming an organo-metallic component, followed by the conversion in the presence of a catalyst complex and a base, preferably in a temperature range from 50 °C to 150 °C under high pressure and the exclusion of oxygen.
  • the compounds of general formula (I), wherein A stands for a group of formula (i) in accordance with the present invention, produced in accordance with the method specified above may have protective groups that can be cleaved, in accordance with the conditions known to one skilled in the art, in order to obtain additional compounds of formula (I).
  • the reaction takes place in inert solvents, in the presence of a dehydrating reagent, optionally in the presence of a base, preferably in a temperature range of -30 °C to 50 °C at normal pressure.
  • subject matter of the present invention is a process for the synthesis of 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1 H-pyrazol-3-yl]-5,6- dihydro-7H-pyrrolo[3,4-d] pyrimidin-7-one (compound A), wherein said process comprises the following starting materials and the synthesis of an acetyl-pyrazyl intermediate; i.e. compound 5. While referring to scheme 1 above, said pre-synthesis comprises the following steps:
  • the general process comprises 11 chemical transformations and follows a linear synthesis approach starting from 3-chloro-5-(trifluoromethoxy)acetophenone (i.e. compound 1 a, see Scheme 10).
  • the second key raw material used is 3-hydrazinepyridine dihydrochloride (i.e. compound 3a; see Scheme 10).
  • 5-(3-chloro-5-(trifluoromethoxy)phenyl)-1-(pyridin-3-yl)-1H-pyrazole-3-carboxylic acid i.e. compound 6a; See Scheme 10) represents the GMP process starting material.
  • 5-(3-chloro-5-(trifluoromethoxy)phenyl)-N-methoxy-N-methyl-1-(pyridin-3-yl)-1H-pyrazole-3-carboxamide represents another common intermediate of the general synthesis process for obtaining 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]-5,6- dihydro-7H-pyrrolo[3,4-d] pyrimidin-7-one (i.e. compound A) in accordance with the invention.
  • Step 1) Starting from the commercially available raw material 3-chloro-5-(trifluoromethoxy)acetophenone (i.e. compound 1 a, see Scheme 10), the chemical transformation towards 5-(3-chloro-5-(trifluoromethoxy)phenyl)-1-(pyridin-3-yl)-1H-pyrazole-3-carboxylic acid (i.e. compound 6a; See Scheme 10) comprises 4 chemical steps, performed as two distinct process steps.
  • Step 1a The first step describes the production and isolation of compound 2a (see Scheme 11) out of compound 1 a, the second process step is designed to produce compound 6a as free acid (see 12).
  • the product compound 2a is isolated in solid form as its potassium salt, and used without further purification in the next process steps (see below).
  • the co-solvent methanol is used for the heredescribed general synthesis in kg-amounts.
  • the presence of methanol improves both the mobility of the obtained suspension, and improves the filtration speed during the isolation of the desired product.
  • Example 1 Further details for the above described first general process step can be derived from the example portion; see Example 1.
  • Step 2 describes the synthesis of compound 6a starting from the intermediate compound 2a (see Scheme 12 below).
  • the reaction as depicted below in Scheme 12 is a two-step single pot reaction.
  • the first step is a condensation reaction of compound 2a with the second key starting material compound 3a.
  • the hydrazone intermediate compound 4a is formed at ambient temperature, but the formation of compound 5a out of compound 4a occurs at elevated temperature.
  • the reaction mixture needs to be heated by slow rate heating.
  • Compound 2a reacts rapidly with compound 3a, resulting in a condensation product, which is not purged in the subsequent reaction steps.
  • the resulting pyridyl-aryl-pyrazole ester compound 5a is a mixture of the methyl and ethyl ester which is not isolated, but converted to the corresponding carboxylic acid compound 6a using an aqueous sodium hydroxide solution.
  • the product compound 6a precipitates out of solution and can be isolated in solid form.
  • the addition of hydrochloric acid solution needs to be monitored on pH, as reaction mixture needs to be of pH ⁇ 2.
  • the obtained dry compound 6a can be used without further purification in the next process step.
  • Step 2a alternatively, compound 6a could also be isolated as either oxalate or hydrochloric acid adduct.
  • the oxalate adduct can be prevented by isolating intermediate compound 2a in solid form by filtration. This filtration step purges all remaining diethyl oxalate, which is the source of the oxalate anion.
  • a hydrochloric acid adduct formation can be prevented.
  • Step 3 This process step describes the synthesis of compound 8a starting from the GMP starting material compound 6a (see Scheme 13 below).
  • the intermediate acid chloride compound 7a is not isolated, but the process is performed as a two-step one pot reaction.
  • the product is isolated in solid form, and used without further purification in the next process step (see below).
  • This reaction is conducted with oxalyl chloride at ambient temperature. Preferably, slow dosing of said reagent oxalyl chloride is performed.
  • the N,O-dimethylhydroxyl amine is added as its HCl salt. No reaction should occur, until this reagent is liberated by dosing of the base.
  • the preferred base with the context of the present invention is diisopropylethylamine (DiPEA).
  • Step 4 This process step 4 describes the synthesis of compound 9a starting from the Weinreb amide compound 8a (see Scheme 14 below).
  • Compound 9a is isolated as a solution in THF. This solution can be used in the next step of the synthesis without further purification.
  • the reagent methylmagnesiumchloride can be commercially obtained as a tetrahydrofuran solution (22 %-wt active material). This solution can be transferred directly from the pressurized container into the reactor.
  • Step 5 Process step 5 is characterized by the synthesis of compound 10a starting from the ketone derivative compound 9a, which is harvested as a tetrahydrofuran solution (see Scheme 15 below).
  • a sodium ethoxide as 21 %-wt solution in ethanol is used for the GMP synthesis of compound 10a .
  • Step 6 The synthesis step 6 describes the synthesis of compound 12a starting from the intermediate compound 10a (see Scheme 16 below).
  • the chemistry is a two-step process in which first a keto-imine (i.e. compound 11 a) is formed, which is not isolated, followed by a Mannich reaction yielding the compound 12a. In both steps the solvent acetic acid is used. The ammonia donor is in both cases ammonium acetate. The paraformaldehyde is dosed as a solid to the reaction mixture, in order to minimize the required amounts of solvents.
  • the first step is performed at approx. 45 ⁇ 5 °C, while the second step is performed at approx. 35 ⁇ 5°C.
  • Dissolution of the process intermediate compound 11 a and the intermediate product compound 10a occurs at temperatures above approx. 30 - 35 °C.
  • the product compound 12a precipitates out of solution by addition of ethanol to the reaction mixture.
  • Step 7 describes the synthesis of compound A as crude product, starting from the intermediate compound 12a (see Scheme 17 below).
  • the reaction mixture is heated and kept under pressure.
  • a positive side effect of performing the reaction under pressure is the fact that the crude isolated compound A is of higher chromatographic purity.
  • Step 7a Purification Isolation and release testing of compound A as crude product:
  • the finalization of the synthesis process is characterized by two distinct purification steps.
  • the first step describes the crystallization of the crude product compound A out of an acetic acid solution, using water as anti-solvent. This step is intended to remove organic impurities formed during the last steps of production, and reduce the amounts of compound 12a, if still present as impurity in the final product.
  • Compound A can be also obtained as acetate adduct. The acetate adduct can be identified by its distinct XRPD pattern. Dosing water, however, yields compound A as a free base with the correct polymorphic form.
  • the second step consists of dissolution of the purified compound A in a mixture of dichloromethane and methanol in order to perform a screening filtration designed to remove any foreign particles, if present.
  • the product can be isolated by filtration after a solvent switch to methanol.
  • Final release testing may also be performed.
  • the compounds of the general formula (I) for use in methods of treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD in accordance with the present invention can act systemically and/or locally.
  • can be administered in a suitable way, such as, e.g., orally, parenterally, pulmonarily, nasally, sublingually, lingually, bucally, rectally, dermally, transdermally, conjunctivally, otically or as an implant or stent.
  • a suitable way such as, e.g., orally, parenterally, pulmonarily, nasally, sublingually, lingually, bucally, rectally, dermally, transdermally, conjunctivally, otically or as an implant or stent.
  • the oral and parenteral administration routes for the compounds of general formula (I) in the methods of treatment and/or prevention of the aforementioned diseases are preferred.
  • the compounds according to the invention can be administered in suitable forms of application.
  • forms of application that release the compounds according to the invention in a quick-acting and/or modified way according to the state of the art and that contain the compounds according to the invention in crystalline and/or amorphized and/or dissolved form and/or in the form of co-crystals or polymorphs of said co-crystals, are selected from a group comprising tablets (uncoated or coated tablets, for example with gastric juice-resistant or slow-dissolving or insoluble coatings, which control the release of the compound according to the invention), tablets or films/wafers that quickly dissolve in the oral cavity, films/lyophilisates, multilayered tablets, capsules (for example, hard- or soft-gelatin capsules), granulates, pellets, powder, emulsions, microemulsions, nanoemulsions, suspensions, nanosuspensions, aerosols or solutions, are suitable.
  • Parenteral administration can be realized by bypassing a resorption step (e.g., by intravenous, intraarterial, intracardial, intraspinal or intralumbar means) or by including resorption (e.g., by intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal means).
  • a resorption step e.g., by intravenous, intraarterial, intracardial, intraspinal or intralumbar means
  • resorption e.g., by intramuscular, subcutaneous, intracutaneous, percutaneous or intraperitoneal means.
  • injection and infusion preparations in the form of solutions, suspensions, nanosuspensions, emulsions, microemulsions, nanoemulsions, lyophilisates, liposomes, polymeric micelles or sterile powders are suitable as forms of application in accordance with the instant invention.
  • inhalation forms of medication inter alia powder inhalers, nebulizers, nose drops, nasal solutions, nasal sprays; e.g. for oral administration tablets that are to be administered lingually, sublingually or buccally; films/wafers or capsules, suppositories, aqueous suspensions (lotions, shaking mixtures), aqueous nanosuspensions, lipophilic suspensions; ear or eye preparations, or e.g. for topical application forms of medication ointments, creams, transdermal therapeutic systems (such as, for example, patches), milk, pastes, foams, scattered powders; implants or stents, are suitable in accordance with the invention.
  • inhalation forms of medication inter alia powder inhalers, nebulizers, nose drops, nasal solutions, nasal sprays; e.g. for oral administration tablets that are to be administered lingually, sublingually or buccally; films/wafers or capsules, suppositories, aque
  • the compounds of the general formula (I) according to the invention can be converted into the above-recited forms of application. This can take place in a way that is known in the art by mixing with inert, nontoxic, pharmaceutically suitable adjuvants.
  • adjuvants include, inter alia, vehicles (for example, microcrystalline cellulose, lactose, mannitol), solvents (e.g., liquid polyethylene glycols), emulsifiers and dispersing agents or wetting agents (for example, sodium dodecyl sulfate, polyoxysorbitanoleate), binders (for example, polyvinylpyrrolidone), synthetic and natural polymers (for example, albumin), stabilizers (e.g., antioxidants, such as, for example, ascorbic acid), dyes (e.g., inorganic pigments, such as, for example, iron oxides), and flavoring and/or odor correctives.
  • vehicles for example, microcrystalline cellulose, lactose, mannitol
  • a single dose preferably contains a compound of general formula (I) as API in quantities from 0.005 to 50 mg/kg, more preferably from 0.005 to 40 mg/kg, even more preferably from 0.05 to 30 mg/kg, even more preferably from 0.05 to 20 mg/kg, even more preferably from 0.05 to 10 mg/kg, most preferred of 0.05 to 5 mg/kg of body weight.
  • a compound of general formula (I) as API in quantities from 0.005 to 50 mg/kg, more preferably from 0.005 to 40 mg/kg, even more preferably from 0.05 to 30 mg/kg, even more preferably from 0.05 to 20 mg/kg, even more preferably from 0.05 to 10 mg/kg, most preferred of 0.05 to 5 mg/kg of body weight.
  • subject matter of the invention are pharmaceutical agents that contain at least one compound of the general formula (I) according to the invention, usually together with one or more inert, non-toxic, pharmaceutically suitable adjuvants, as well as their use for the above-mentioned purposes.
  • the present invention is directed to pharmaceutical combination compositions comprising:
  • compositions comprising at least one compound of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal, and one or more other active ingredients, in particular for its use in methods of treatment and/or prevention of the aforementioned diseases.
  • suitable combination active ingredients comprising:
  • Agents having antithrombotic activity preferably mean compounds selected from the group of platelet aggregation inhibitors, anticoagulants or fibrinolytic agents.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal are to be administered in a method of treatment and/or prevention of the aforementioned diseases/conditions in combination with a platelet aggregation inhibitor, being selected for example and preferably from a group comprising aspirin, clopidogrel, ticlopidine or dipyridamole.
  • antihypertensive agents used in combination with the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal in a method of treatment and/or prevention of the aforementioned diseases/conditions are preferably compounds selected from the group comprising calcium antagonists, angiotensin A-II antagonists, ACE inhibitors, endothelin antagonists, renin inhibitors, alpha-receptor blockers, beta-receptor blockers, mineralocorticoid receptor antagonists, and of diuretics.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with a calcium antagonist such as by way of example and preferably selected from a group comprising nifedipine, amlodipine, verapamil or diltiazem.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with an alpha 1 receptor blocker such as by way of example and preferably prazosin.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with a beta-receptor blocker, such as for example and not being limited to, propranolol, atenolol, timolol, pindolol, alprenolol, oxprenolol, penbutolol, bupranolol, metipranolol, nadolol, mepindolol, Carazalol, sotalol, metoprolol, betaxolol, celiprolol, bisoprolol, carteolol, esmolol, labetalol, carvedil
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with an angiotensin A-II antagonist such as, for example and preferably selected from a group comprising losartan, candesartan, valsartan, telmisartan or embursatan.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with an ACE inhibitor such as by way of example and preferably enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • an ACE inhibitor such as by way of example and preferably enalapril, captopril, lisinopril, ramipril, delapril, fosinopril, quinopril, perindopril or trandopril.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with an endothelin antagonist, such as for example and preferably selected from a group comprising bosentan, darusentan, ambrisentan or sitaxsentan.
  • lipid metabolism-modifying agents are preferably compounds selected from the group comprising CETP inhibitors, thyroid receptor agonists, cholesterol synthesis inhibitors such as HMG-CoA reductase or squalene synthesis inhibitors, ACAT inhibitors, MTP inhibitors, PPAR-alpha, PPAR-gamma and/or PPAR-delta agonists, cholesterol absorption inhibitors, polymeric bile acid reabsorption inhibitors, lipase inhibitors and lipoprotein antagonists.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with a cholesterol absorption inhibitor such as, for example and preferably selected from a group comprising ezetimibe, tiqueside or pamaqueside.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with a lipase inhibitor such as, for example and preferably orlistat.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for use in a method of treatment and/or prevention of the aforementioned diseases/conditions are to be combined with a polymeric bile as for example and preferably selected from a group comprising cholestyramine, colestipol, colesolvam, CholestaGel or colestimid.
  • compositions comprising: a therapeutically effective amount of a composition comprising
  • lipid modulating agents include a lipase inhibitor, an HMG-CoA reductase inhibitor, an HMG-CoA synthase inhibitor, an HMG-CoA reductase gene expression inhibitor, an HMG-CoA synthase gene expression inhibitor, an MTP/Apo B secretion inhibitor, a CETP inhibitor, a bile acid absorption inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a squalene synthetase inhibitor, a squalene epoxidase inhibitor, a squalene cyclase inhibitor, a combined squalene epoxidase/squalene cyclase inhibitor, a fibrate, niacin, a combination of niacin and lovastatin, an ion-exchange resin, an antioxidant, an ACAT inhibitor and a bile acid sequestrant.
  • a lipase inhibitor an HMG-CoA reduct
  • the compounds of the invention may be used in the herein dislcosed methods for treatment and/or prophylaxis of the aforementioned diseases alone or in combination, if necessary, with other agents.
  • Said other agents may be preferably selected from the classes of statins, prile, beta blocker, acetylsalicylic acid, sartanes, ACE-inhibitor, AT-1 receptor antagonists, diuretics, calcium-channel antagonists/calcium-channel blocker.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal may also be used in conjunction with other pharmaceutical agents (e.g., LDL-cholesterol lowering agents, triglyceride lowering agents) in the methods of treatment and/or prevention as described herein.
  • other pharmaceutical agents e.g., LDL-cholesterol lowering agents, triglyceride lowering agents
  • they may be used in combination with lipid modulating agents, antidiabetic agents and further cardiovascular agents.
  • Lipid modulating agents may be used as a combination agent in conjunction with compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal.
  • Any HMG-CoA reductase inhibitor may be used in the combination aspect of this invention.
  • the conversion of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) to mevalonate is an early and rate-limiting step in the cholesterol biosynthetic pathway. This step is catalyzed by the enzyme HMG-CoA reductase.
  • Statins inhibit HMG-CoA reductase from catalyzing this conversion. The following paragraphs describe exemplary stati ns.
  • HMG-CoA reductase inhibitor refers to compounds, which inhibit the bioconversion of hydroxymethylglutaryl-coenzyme A to mevalonic acid catalyzed by the enzyme HMG-CoA reductase. Such inhibition is readily determined by those skilled in the art according to standard assays (e.g., Meth. Enzymol. 1981; 71:455-509 and references cited therein). A variety of these compounds are described and referenced below, however, other HMG-CoA reductase inhibitors will be known to those skilled in the art.
  • U.S. Pat. No. 4,231,938 describes certain compounds isolated after cultivation of a microorganism belonging to the genus Aspergillus, such as lovastatin.
  • U.S. Pat. No. 4,444,784 describes synthetic derivatives of the aforementioned compounds, such as simvastatin. Also, U.S. Pat. No. 4,739,073 describes certain substituted indoles, such as fluvastatin. Also, U.S. Pat. No. 4,346,227 describes ML-236B derivatives, such as pravastatin. Also, EP-491226A describes certain pyridyldihydroxyheptenoic acids, such as cerivastatin. In addition, U.S. Pat. No.
  • Atorvastatin calcium i.e., atorvastatin hemicalcium
  • Lipitor ® a trademark of Lipitor ®
  • Statins also include such compounds as rosuvastatin described in U.S. RE37,314 E , pitivastatin described in EP 304063 B1 and U.S. Pat. No. 5,011,930 , simvastatin, described in U.S. Pat. No. 4,444,784 ; pravastatin, described in U.S. Pat. No. 4,346,227 ; cerivastatin, described in U.S. Pat. No. 5,502,199 ; mevastatin, described in U.S. Pat. No. 3,983,140 ; velostatin, described in U.S. Pat. No. 4,448,784 and U.S. Pat. No.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal may be administered with antidiabetic compounds in methods of treating and/or preventing the comorbidity diabetes.
  • Diabetes can be treated by administering to a patient having diabetes (especially Type II), insulin resistance, impaired glucose tolerance, metabolic syndrome, or the like, or any of the diabetic complications such as neuropathy, nephropathy, retinopathy or cataracts, a therapeutically effective amount of a compound of the present invention in combination with other agents (e.g., insulin) that can be used to treat diabetes.
  • a therapeutically effective amount of a compound of the present invention in combination with other agents e.g., insulin
  • any glycogen phosphorylase inhibitor can be used as the second agent in combination with a compound of the present invention or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal.
  • glycogen phosphorylase inhibitor refers to compounds that inhibit the bioconversion of glycogen to glucose-1-phosphate, which is catalyzed by the enzyme glycogen phosphorylase. Such glycogen phosphorylase inhibition activity is readily determined by those skilled in the art according to standard assays (e.g., J. Med. Chem. 41 (1998) 2934-2938 ). A variety of glycogen phosphorylase inhibitors are known to those skilled in the art including those described in WO 96/39384 and WO 96/39385 .
  • any aldose reductase inhibitor can be used in combination with a compound of the present invention.
  • aldose reductase inhibitor refers to compounds that inhibit the bioconversion of glucose to sorbitol, which is catalyzed by the enzyme aldose reductase.
  • Aldose reductase inhibition is readily determined by those skilled in the art according to standard assays (e.g., J. Malone, Diabetes, 29:861-864 (1980). "Red Cell Sorbitol, an Indicator of Diabetic Control ").
  • a variety of aldose reductase inhibitors are known to those skilled in the art, such as those described in U.S. Pat. No. 6,579,879 , which includes 6-(5-chloro-3-methyl-benzofuran-2-sulfonyl)-2H-pyridazin-3-one.
  • any sorbitol dehydrogenase inhibitor can be used in combination with a compound of the present invention or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal.
  • sorbitol dehydrogenase inhibitor refers to compounds that inhibit the bioconversion of sorbitol to fructose, which is catalyzed by the enzyme sorbitol dehydrogenase.
  • Such sorbitol dehydrogenase inhibitor activity is readily determined by those skilled in the art according to standard assays (e.g., Analyt.
  • sorbitol dehydrogenase inhibitors are known, for example, U.S. Pat. Nos. 5,728,704 and 5,866,578 describe compounds and a method for treating or preventing diabetic complications by inhibiting the enzyme sorbitol dehydrogenase.
  • calcium channel blockers which are within the scope of the present invention include, but are not limited to: bepridil, which may be prepared as described in U.S. Pat. No. 3,962, 238 or U.S. Reissue No. 30,577 ; clentiazem, which may be prepared as described in U.S. Pat. No. 4,567,175 ; diltiazem, which may be prepared as described in U.S. Pat. No. 3,562 , fendiline, which may be prepared as described in U.S. Pat. No. 3,262,977 ; gallopamil, which may be prepared as described in U.S. Pat. No.
  • mibefradil which may be prepared as described in U.S. Pat. No. 4,808,605 ; prenylamine, which may be prepared as described in U.S. Pat. No. 3,152,173 ; semotiadil, which may be prepared as described in U.S. Pat. No. 4,786,635 ; terodiline, which may be prepared as described in U.S. Pat. No. 3,371,014 ; verapamil, which may be prepared as described in U.S. Pat. No. 3,261,859 ; aranipine, which may be prepared as described in U.S. Pat. No. 4,572,909 ; barnidipine, which may be prepared as described in U.S.
  • nimodipine which may be prepared as described in U.S. Pat. No. 3,799,934
  • nisoldipine which may be prepared as described in U.S. Pat. No. 4,154,839
  • nitrendipine which may be prepared as described in U.S. Pat. No. 3,799,934
  • cinnarizine which may be prepared as described in U.S. Pat. No. 2,882,271
  • flunarizine which may be prepared as described in U.S. Pat. No. 3,773,939
  • lidoflazine which may be prepared as described in U.S. Pat. No.
  • Examples of presently marketed products containing antihypertensive agents include calcium channel blockers, such as Cardizem ® , Adalat ® , Calan ® , Cardene ® , Covera ® , Dilacor ® , DynaCirc ® , Procardia XL ® , Sular ® , Tiazac ® , Vascor ® , Verelan ® , Isoptin ® , Nimotop ® , Norvasc ® , and Plendil ® ; angiotensin converting enzyme (ACE) inhibitors, such as Accupril ® , Altace ® , Captopril ® , Lotensin ® , Mavik ® , Monopril ® , Prinivil ® , Univasc ® , Vasotec ® and Zestril ® .
  • calcium channel blockers such as Cardizem ® , Adalat ® , Calan ® ,
  • angiotensin converting enzyme inhibitors which are within the scope of the present invention include, but are not limited to: alacepril, which may be prepared as described in U.S. Pat. No. 4,248,883 ; benazepril, which may be prepared as described in U.S. Pat. No. 4,410,520 ; captopril, which may be prepared as described in U.S. Pat. Nos. 4,046,889 and 4,105,776 ; ceronapril, which may be prepared as described in U.S. Pat. No. 4,452,790 ; delapril, which may be prepared as described in U.S. Pat. No.
  • quinapril which may be prepared as described in U.S. Pat. No. 4,344,949 ; ramipril, which may be prepared as described in U.S. Pat. No. 4,587,258 ; spirapril, which may be prepared as described in U.S. Pat. No. 4,470,972 ; temocapril, which may be prepared as described in U.S. Pat. No. 4,699,905 ; and trandolapril, which may be prepared as described in U.S. Pat. No. 4,933,361 .
  • angiotensin-II receptor antagonists which are within the scope of the present invention include, but are not limited to: candesartan, which may be prepared as described in U.S. Pat. No. 5,196,444 ; eprosartan, which may be prepared as described in U.S. Pat. No. 5,185,351 ; irbesartan, which may be prepared as described in U.S. Pat. No. 5,270,317 ; losartan, which may be prepared as described in U.S. Pat. No. 5,138,069 ; and valsartan, which may be prepared as described in U.S. Pat. No. 5,399,578 .
  • beta-adrenergic receptor blockers which are within the scope of the present invention include, but are not limited to: acebutolol, which may be prepared as described in U.S. Pat. No. 3,857,952 ; alprenolol, which may be prepared as described in Netherlands Patent Application No. 6,605,692 ; amosulalol, which may be prepared as described in U.S. Pat. No. 4,217,305 ; arotinolol, which may be prepared as described in U.S. Pat. No. 3,932,400 ; atenolol, which may be prepared as described in U.S. Pat. No.
  • bufetolol which may be prepared as described in U.S. Pat. No. 3,723,476
  • bufuralol which may be prepared as described in U.S. Pat. No. 3,929,836
  • bunitrolol which may be prepared as described in U.S. Pat. Nos. 3,940,489 and 3,961,071
  • buprandolol which may be prepared as described in U.S. Pat. No. 3,309,406
  • butiridine hydrochloride which may be prepared as described in French Patent No. 1,390,056
  • butofilolol which may be prepared as described in U.S. Pat. No.
  • carazolol which may be prepared as described in German Patent No. 2,240,599 ; carteolol, which may be prepared as described in U.S. Pat. No. 3,910,924 ; carvedilol, which may be prepared as described in U.S. Pat. No. 4,503,067 ; celiprolol, which may be prepared as described in U.S. Pat. No. 4,034,009 ; cetamolol, which may be prepared as described in U.S. Pat. No. 4,059,622 ; cloranolol, which may be prepared as described in German Patent No.
  • metipranolol which may be prepared as described in Czechoslovakian Patent Application No. 128,471 ; metoprolol, which may be prepared as described in U.S. Pat. No. 3,873,600 ; moprolol, which may be prepared as described in U.S. Pat. No. 3,501,7691 ; nadolol, which may be prepared as described in U.S. Pat. No. 3,935,267 ; nadoxolol, which may be prepared as described in U.S. Pat. No. 3,819,702 ; nebivalol, which may be prepared as described in U.S. Pat. No.
  • the present invention is further directed to the use of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal as a medicament (such as a unit dosage tablet or unit dosage capsule, or a dual, tripple or polypill with at least one of the additional pharmaceutical agents as recited herein) in a method of treatment and/or prevention of one or more of the conditions previously identified in the above sections discussing its use in methods of treatment and/or prevention.
  • a medicament such as a unit dosage tablet or unit dosage capsule, or a dual, tripple or polypill with at least one of the additional pharmaceutical agents as recited herein
  • a “polypill” is a medication that is a drug product in pill form (i.e., tablet or capsule) that combines multiple active pharmaceutical ingredients.
  • the prefix “poly” means “multiple,” referring to the multiplicity of distinct drugs in a given "pill.”
  • An occasional synonym with the context of the invention is combopill.
  • Polypills may be administered in methods of treatment/prevention of the aforementioned diseases and / or comorbidities of the invention as a means of preventive medicine, and/or treating actual pathophysiological condition(s), the former typically involving lower dosages than the latter.
  • polypills may reduce the number of tablets or capsules (e.g. when orally administered) that need to be taken, which in turn may facilitate handling and administration of pharmaceuticals as well as alleviate patient pill-burden.
  • the multiple drugs in a given polypill might all be aimed at a single underlying condition (or, group of related conditions).
  • polypill(s) is/are a combinatorial drug product(s) of the aforemention pharmaceutical drug combinations.
  • polypills can also be individually custom-made for specific patients through so-called pharmacy compounding.
  • the present invention comprises the use of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal for the manufacture of a medicament (such as a unit dosage tablet or unit dosage capsule, or a dual, tripple or polypill with at least one of the additional pharmaceutical agents as recited herein) for use in a method of treatment and/or prevention of one or more of the conditions previously identified in the above sections discussing its use in methods of treatment and/or prevention.
  • a medicament such as a unit dosage tablet or unit dosage capsule, or a dual, tripple or polypill with at least one of the additional pharmaceutical agents as recited herein
  • the present invention is further directed to pharmaceutical compositions of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate thereof, particularly a hydrate thereof, or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal.
  • a pharmaceutical composition may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses.
  • a " unit dose" is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient; which is at least a compound of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal of the invention.
  • the amount of said active ingredient is generally equal to the dosage of the active ingredient, which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • composition of the invention may be prepared, packaged, or sold in form of a dual, tripple or polypill with one or more of the additional pharmaceutical agents as recited above.
  • the amount of the active ingredient is also generally equal to the dosage of the active ingredient, which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • the present invention is further directed to advantageous pharmaceutical formulations of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal.
  • the inventors surprisingly and unexpectedly found specific pharmaceutical formuliations that significantly increase solubility of the compounds of the general formula (I), and particularly of the compounds A and C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal, which results in an advatageous dissolution behavior and thus increased bioavailability after being administered in the methods of treatment and/or prevention of the aforementioned diseases/conditions and / or its comorbidities.
  • compound A is a synthetic heterocyclic compound with molecular weight around 500 and pKa at 2.3. Typically, this compound exhibits very poor solubility in aqueous media (pH 2.0: 0.4 mg/ml, pH 7.4: 50 - 70 ⁇ g/ml). Also, compound A shows very low solubility in the commonly used pharmaceutical excipients, such as oils, cosolvents, and surfactants. But compound A is stable in aqueous and physiological fluids as well as organic solvents.
  • the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal may be administered as a pharmaceutical formulation comprising a pharmaceutically effective amount of a compound of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal in association with one or more pharmaceutically acceptable excipients including carriers, vehicles and diluents in the methods of treatment/prevention as disclosed herein.
  • the compounds as disclosed herein may be formulated for oral, buccal, intranasal, parenteral (e.g., intravenous (i.v.), intramuscular (i.m.) or subcutaneous (s.c.)), transdermal or rectal administration or in a form suitable for administration by inhalation.
  • parenteral e.g., intravenous (i.v.), intramuscular (i.m.) or subcutaneous (s.c.)
  • transdermal or rectal administration e.g., transdermal or rectal administration or in a form suitable for administration by inhalation.
  • the compounds of the invention may also be formulated for sustained delivery.
  • the inventors found the use of hot-melt extrusion suitable to formulate the compounds of the general formula (I), particularly of compounds A and C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal as poorly water-soluble API S .
  • the inventors found the following solid dispersions to improve bioavailability of the poorly soluble drugs by converting a crystalline API into its corresponding amorphous form.
  • solid dispersion formulations for the compounds of general formula (I), and particularly for the compounds A and C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal are provided that are based on
  • the hot-melt extrusion process was able to fully convert crystalline API; i.e. a compound of general formula (I), particularly a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal into its corresponding amorphous form for the following pharmaceutical formulation of the invention:
  • a stable solid dispersion of the compounds of general formula (I), particularly of compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal is obtainable by using the following ranges of API and relevant excipients:
  • preferred polymers for preparing said solid dispersion of a compound of general formula (I), particularly of compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal are selected from a group comprising celluloses / modified cellulose, Eudragits and polyvinyl pyrrolides, poloxamers, and PEGs.
  • preferred organic acids that are to be added to improve solubilization of compounds of general formula (I), particularly of compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal in the polymeric matrix are selected from a group comprising citric acid, maleic acid, tartaric acid, ascorbic acid, adipic acid, aspartatic acid, benzenesulfonic acid, glucoheptonic acid, D-gluconic acid, L-glutamic acid, lactic acid, L-lysine, methanesulfonic acid, p-toluenesulfonic acid.
  • nanomilling allows to formulate a stable nanoparticular suspension with a compound of general formula (I), particularly of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal and so to decrease the particle size, whereby simultaneously the particle surface increases significantly, which improves bioavailability of the compound of general formula (I), particularly of compound A or C or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal as API.
  • nanosuspension formulations comprising:
  • both above mentioned surfactants and polymers represent stabilizers for the herein disclosed nanosuspension formulations of a compound of general formula (I), particularly of a compound A or C or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal.
  • a preferred nanosuspension comprises at least one compound of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compound or a pharmaceutically acceptable co-crystal of said compound or a polymorph of said co-crystal and Sodiumglycocholate (SGC), Poloxamer 188, and water.
  • SGC Sodiumglycocholate
  • Another preferred nanosuspension of the invention comprises at least one compound of the general formula (I), particularly of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal and Sodiumdeoxycholate (SDC), Povidone (PVP K25) and water.
  • SDC Sodiumdeoxycholate
  • PVP K25 Povidone
  • the above disclosed nanosuspensions are provided as formulations for the compounds of the general formula (I), particularly for the compounds A and C or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal.
  • the here disclosed stable nanosuspensions of the compounds of general formula (I), particularly of the compounds A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal are provided for a long acting parenteral, for subcutaneous, for transdermal as well as for oral application thereof in the the methods of treatment/prevention for the herein disclosed cardiovascular diseases and / or comorbidities thereof.
  • the inventors found a mesoporous silica particle based formulation of the compounds of general formula (I), in particular of a compound A or C or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal as API to convert the crystalline API into the corresponding amorphous form, which surprisingly and unexpectedly increases dissolution rate of the API and thus its bioavailability.
  • Mesoporous silica particles have typical diameter in the range of approx. 50-250 ⁇ m and a narrow particle size distribution.
  • the structure and morphology of these particles can be controlled at nano to micro scale and enable to generate high surface areas and pore volumes allowing for high drug loading; e.g. of 40 wt % - 50 wt % API load.
  • the entrapment of drugs in the mesopores leads to conversion of crystalline drug into its amorphous form.
  • the instant inventors provide a stable mesoporous silica particle based formulation of the compounds of general formula (I), in particular of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal which is obtainable by impregnation of approx.
  • a compound of general formula (I) as API particularly of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal onto silica material, preferably OMS-7 silica material.
  • the concentration of silica material in the pharmaceutical formulations based on mesoporous silica particles can range from 10 wt % to 95 wt %.
  • a compound of general formula (I) particularly of a compound A or C or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal of the invention as API from the mesoporous silica material
  • various polymers and surfactants can be added to the above formulation.
  • the concentration of said polymers and surfactants may range from 0.01 wt % to 5 wt %.
  • said surfactants are preferably selected from a group comprising polysorbates, spans, poloxmaers, TPGS, lecithin.
  • said polymers are preferably selected from a group comprising celluloses, polyvinylpyrrolidones (PVP), polythethylene glycols (PEG), Soluplus.
  • said surfactants and polymers can range in concentration between 1 wt % and 40 wt %, provided that a 1:1 ratio with the compound of general formula (I), particularly of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal is given.
  • the inventors surprisingly found that sodium lauryl sulphate (SLS) is advantageous as surfactant and that hydroxypropylmethyl cellulose (HPMC) is advantageous as polymer.
  • SLS sodium lauryl sulphate
  • HPMC hydroxypropylmethyl cellulose
  • the ratio of said surfactant and polymer with a compound of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal as API is 1:1 in accordance with the present invention.
  • the here disclosed silica-based drug delivery technology for the compounds of general formula (I), particularly for a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal provides for a solubility-enhancing formulation for the otherwise poorly water-soluble compounds of general formula (I), particularly compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal as API of the invention.
  • Capmul ® -MCM EP C8 based formulations of the compounds of general formula (I), particulary of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal are disclosed, said Capmul ® -based formulations comprising:
  • the inventors further found that for the compounds of the invention in higher concentrations of ⁇ 200 mg/kg, preferably ⁇ 300 mg/kg, more preferably ⁇ 400 mg/kg, most preferred ⁇ 500 mg/kg, suspension formulation with the vehicle Capmul ® allows for suitable administration thereof; whereby the Labrafil concentration is not higher than 50 %.
  • the suspension formulations of the present invention allowed to administer compounds of general formula (I), particularly of a compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal with doses up to 500 mg/kg - without the need of a concentration of Labrafil above 50 %.
  • the drug loading of a compound of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal can range from 0.1 to 50 mg/ml and the Capmul MCM EP C8 concentration can range from 5% to 70%, whereby the acetic acid component in this formulation can range from 1% to 15%, and the Labrafil ® component can range from 5% to 50%.
  • the present invention has an aspect that relates to the use of the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in methods of treatment and/or prevention of the disease/conditions described herein with a combination of active ingredients which may be administered separately (see above), the invention also relates to combining separate pharmaceutical compositions in form of a kit.
  • the kit of the invention comprises at least two separate pharmaceutical compositions: a compound of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal and a second compound as described above.
  • the kit of the invention further comprises a means for containing the separate compositions such as a container, a divided bottle or a divided foil packet.
  • a means for containing the separate compositions such as a container, a divided bottle or a divided foil packet.
  • the kit comprises directions for the administration of the separate components.
  • kits may comprise bilayered tablets sachet pellets filled in capsules (i.e. the so-called stick-pack technology).
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral, parenteral, long acting parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
  • different dosage forms e.g., oral, parenteral, long acting parenteral
  • the present invention has an aspect that relates to the use of the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in methods of treatment and/or prevention of the disease/conditions described herein with a combination of active ingredients which may be administered jointly, the invention also relates to combining separate pharmaceutical compositions in a single dosage form, such as (but not limited to) a single tablet or capsule, a bilayer or multilayer tablet or capsule, or through the use of segregated components or compartments within a tablet or capsule.
  • the present invention has an aspect that relates to the use of the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in methods of treatment and/or prevention of the disease/conditions described herein with a combination of one or more active ingredients which may be administered jointly, the invention also relates to combining separate pharmaceutical compositions in a dual, tripple or polypill dosage form.
  • the active ingredient of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in accordance with the invention may be delivered as a suspension or a nanosuspension in an aqueous or non-aqueous vehicle, with or without additional solvents, co-solvents, excipients, or complexation agents selected from pharmaceutically acceptable diluents, excipients, vehicles, or carriers.
  • compound A was orally administered as:
  • Compound A was formulated as suspension in Labrafil ® M1944CS, mesoporous silica particles, aqueous nanosuspension and co-solvent based solution. Single doses of 30 or 500 mg/kg were administered to mice except the co-solvent based solution, which was administered at 100 or 500 mg/kg.
  • a single oral dose of 30 or 100 mg/kg was applied to dogs. Pharmacokinetic parameters were assessed and compared (see Example 7). Thereby, micronized, non-micronized compound A and the ethane sulfonate of compound A were orally administered to dogs formulated as a Labrafil ® M1944CS suspension at a dose of 30 mg/kg. The micronization of compound A and the use of an ethane sulfonate of compound A showed no increase of exposure when compared to the non-micronized material.
  • compound A administered as Labrafil ® M1944CS suspension and as mesoporous silica particles showed the most favourable results in terms of C max and maximal exposure in both species; dogs and mice. Similar plasma concentration time profiles were observed after oral administration of compound A formulated either as Labrafil ® M1944CS suspension or as mesoporous silica particles.
  • the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal may be administered as Labrafil ® M1944CS suspension and as mesoporous silica particles formulation to achieve an improved pharmacokintetic spectrum in an individual who is in need of such administration.
  • the compounds of the general formula (I), and particularly the compound A or C, or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal are therefore suitable for use as medicaments in a method for treating and/or preventing cardiovascular diseases and / or comorbidities thereof, in particular for use in methods of treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD in humans and animals.
  • the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal of the invention are suitable for the use in methods of treatment and/or prevention of the aforementioned diseases.
  • the compounds of the invention can therefore be also used in medicaments for the treatment and/or prevention of cardiovascular diseases such as high blood pressure (hypertension), resistant hypertension, acute and chronic heart failure, coronary heart disease, stable and unstable angina pectoris, peripheral and cardiac vascular disorders, arrhythmias, arrhythmia of the atria and the chambers and conduction disorders such as atrio-ventricular blockade grade I-III (AV block I-III), supraventricular tachyarrhythmia, atrial fibrillation, atrial fibrillation, ventricular fibrillation, ventricular flutter, ventricular tachyarrhythmia, torsades de pointes tachycardia, extrasystoles of atrium and the ventricle, AV junctional premature beats, sick sinus syndrome, syncope, Wolff-Parkinson-White syndrome, of acute coronary syndrome (ACS), autoimmune heart disease (pericarditis, endocarditis, Valvolitis, a
  • the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal of the invention are provided for the use in methods of treatment and / or prevention of cardiovascular comorbidities as atherosclerosis, lipid metabolism disorders, hypolipoproteinaemias, dyslipidemia, hypertriglyceridemia, hyperlipidemia, hypercholesterolaemia, abetelipoproteinaemia, sitosterolaemia, cholesterosis, Tangier disease, obesity (adiposity), and combined hyperlipidemia and the metabolic syndrome.
  • the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal of the invention are provided for the use in methods of the treatment and / or prevention of primary and secondary Raynaud's phenomenon, of microcirculation impairments, claudication, peripheral and autonomic neuropathy, diabetic microangiopathy, diabetic retinopathy, diabetic ulcers on the extremities, gangrene, CREST syndrome, erythematosis, onychomycosis, rheumatic diseases, and may be used to promote wound healing.
  • the compounds of the present invention are further characterised particularly by an advantageous HCN4 inhibition profile characterized by an IC 50 in vitro of below 1 ⁇ M, preferably below 0.8 ⁇ M, more preferably below 0.6 ⁇ M, even more preferably below 0.5 ⁇ M when determined in CHO cells transiently expressing the HCN4 channel by a suitable IC 50 measurement.
  • additional subject matter of the present invention is therefore the use of the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in accordance with the invention in a method for treating and/or preventing cardiovascular diseases and/or comorbidties thereof in a subject suffering therefrom, wherein said subject is characterized by having a resting daytime sinus rate of ⁇ 90 beats/min. (bpm), preferably ⁇ 100 beats/min. (bpm) and an average 24-hours heart rate of ⁇ 90 beats/min. (bpm) when measured by e.g. Holter monitoring or any other suitable method the person skilled in the art is aware of that are otherwise ruled out for being related to physiologic demands or conditions known to increase heart rate.
  • a preferred subject matter of the present invention is the use of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in a method for treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD.
  • cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke
  • cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke
  • cardiovascular diseases selected from the group comprising CA, HHD,
  • Yet a more preferred subject matter of the present invention is the use of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in accordance with the invention in a method for treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, IST, SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, and obesity.
  • cardiovascular diseases selected from the group comprising CA, HHD, IST, SAP, MI, CAD, HF, and stroke
  • a most preferred subject matter of the present invention is the use of the compounds of the general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in accordance with the invention in a method for treating and/or preventing cardiovascular diseases selected from the group comprising IST, SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes and metabolic syndrome.
  • subject matter of the present invention is a method for treating and/or preventing cardiovascular diseases selected from the group comprising CA, HHD, tachycardia, IST, AP and SAP, MI, CAD, HF, and stroke, and/or for use in methods of treating and/or preventing cardiovascular comorbidities selected from the group comprising diabetes, metabolic syndrome, obesity, and CKD using a therapeutically effective amount of the compounds in accordance with the present invention or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal.
  • an additional subject matter of the present invention is, in particular, the use of a therapeutically effective quantity of the compounds of general formula (I) or a pharmaceutically acceptable salt or solvate or a solvate of the salt of said compounds or a pharmaceutically acceptable co-crystal of said compounds or a polymorph of said co-crystal in accordance with the invention in a method to prevent and/or treat cardiovascular diseases caused by increased heart rates and/or heart arrhythmias and/or hypertension.
  • AUC 0-24 Area under the analyte vs. time concentration curve from time of administration up to 24h post dosing AUC last Area under the analyte vs. time concentration curve from time of administration up to the time of the last quantifiable concentration post dosing
  • AUC 0- ⁇ Area under the analyte vs. time concentration curve from time of administration up to infinity AUC 0- ⁇ /D Dose normalized area under the analyte vs.
  • Example 1 Synthesis process for obtaining 4-[5-[3-Chloro-5-(trifluoromethoxy) phenyl]-1-(pyridin-3-yl)-1H-pyrazol-3-yl]-5,6-dihydro-7H-pyrrolo[3,4-d]pyrimidin-7-one (i.e. compound A)
  • Step 2) Synthesis of a compound 6a out of compound 2a
  • a very slow stirring speed needs to be applied due to the fact that ammonium acetate has a tendency to form large chunks of solids, which may damage the equipment. Dissolution of the reagent occurs upon heating. When full dissolution is obtained, the stirrer speed is increased to a medium speed. When full dissolution is observed the conversion from compound 10a towards compound 11 a is also nearly completed.
  • Step 7a Purification isolation and release testing of compound A as crude product
  • RX-1 450 L glass line reactor.
  • RX-2 1000 L glass lined reactor.
  • FD 200 L Hastelloy filter dryer.
  • Example 1 The analytical results for an exemplary batch synthesized by the above-described Example 1 can be dervided from FIG. 5 (i.e. final release testing).
  • Vehicle control (0.1% DMSO) and ZD7288 (100 ⁇ M), a suitable blocker of the HCN4 channel, were used as additional controls to assure accuracy of the method used.
  • the whole-cell patch-clamp technique was used to demonstrate the effects of the six tested compounds of the invention in direct comparison to the above-mentioned reference compounds Ivabradine, and the compounds X and Y on HCN4 steady state currents.
  • the compounds were tested first at one concentration of 10 ⁇ M (compound A, compound Y, compound X, compound B, compound E, compound F and ivabradine) or 1 ⁇ M for (compound C and D). If the current blocking of HCN4 at this concentration was higher than 50% the compounds were tested again at five concentrations of a half-logarithmic dilution series starting with either 10 ⁇ M or 1 ⁇ M as mentioned above (meaning 0.1 ⁇ M, 0.3 ⁇ M, 1 ⁇ M, 3 ⁇ M, and 10 ⁇ M or 0.01 ⁇ M, 0.03 ⁇ M 0.1 ⁇ M, 0.3 ⁇ M and 1 ⁇ M).
  • the above described dose levels of the tested compounds were applied as follows. The tested compounds were weighed into a container on a calibrated precision balance. Vehicle was added to achieve a clear stock solution without precipitated test compounds. Dependending on compound solubility a stock solution of 10 mM or 1 mM (Compound C and D) was initially prepared (storage at 19 °C to 30 °C).
  • the 1x bath solution was prepared by diluting 10x bath solution without glucose and 100x glucose solution with water at least every 7 days. Both stock solutions had been prepared prior to the experimental start and stored at 1°C to 9°C (10x bath solution) or -10 °C to -30 °C (100x glucose solution). When in use, the 1x bath solution was kept at room temperature (19 °C - 30 °C). When not in use, the 1x bath solution was stored at 1 °C to 9 °C.
  • the final bath solution included the components: sodium chloride 137 mM; potassium chloride 4.0 mM; calcium chloride 1.8 mM; magnesium chloride 1.0 mM; HEPES 10 mM; D-Glucose 10 mM; pH (NaOH) 7.4.
  • the 1 x pipette solutions were thawed every day out of a frozen 1 x pipette solution, which had been prepared prior to the experimental start, aliquoted and stored at - 10 °C to - 30 °C.
  • the 1x pipette solutions were filled into the patch-pipettes and kept at room temperature (19°C - 30°C).
  • the 1x pipette solutions included the components outlined below: cesium fluoride 135 mM; sodium chloride 10 mM; calcium chloride 0.2 mM; HEPES 10 mM; EGTA 5 mM; cAMP 10 mM; pH (CsOH) 7.3.
  • Test system CHO cells transiently expressing the HCN4 channel; NCBI Reference Sequence: NP005468.1 (HCN4 channel); Source CHO: ATCC, CCL-61 LGC Standards GmbH, Wesel, Germany were used.
  • Equipment Amplifier EPC-9/10, HEKA Electronics
  • Headstage Preamplifier EPC-9/10, HEKA Electronics
  • Software Patchmaster HEKA Electronics.
  • CHO cells were passaged at a confluence of about 50 - 80 %.
  • cells were seeded onto 35 mm sterile culture dishes containing 2 mL culture complete medium without antibiotics. Cells were cultivated at a density that enables the recording of single cells (without visible connections to other cells) to avoid electrical coupling.
  • CHO cells were seeded at a density allowing single cells to be recorded on the dish holder of the microscope and continuously perfused (at approximately 1 mL/min) with the bath solution described above. All solutions applied to cells including the pipette solution were maintained at room temperature (19 °C - 30 °C). After formation of a Gigaohm seal between the patch electrodes and individual cells (pipette resistance range: 2.0 M ⁇ - 7.0 M ⁇ ; seal resistance range: > 1 G ⁇ ) the cell membrane across the pipette tip was ruptured to assure electrical access to the cell interior (whole-cell patch-configuration).
  • SigmaPlot 11 was used to calculate the means ⁇ SEM of relative steady state current blockade for each concentration of the tested compounds. Analysis of Variance (ANOVA), multisample comparison (Dunnett) was conducted to test statistical significance of the test concentrations and the reference compounds versus vehicle (0.1 % DMSO in bath solution) using GraphPad Prism 5. Representative currents determined for the tested compounds in CHO cells are given in the Figures 6 to 9 .
  • X is the actual applied drug concentration
  • IC 50 is the concentration of drug at half maximal inhibition
  • H the Hill coefficient.
  • the "Hill coefficient” provides a way to quantify this effect. It describes the fraction of the macromolecule saturated by ligand as a function of the ligand concentration; it is used in determining the degree of cooperativeness of the ligand binding to the enzyme or receptor.
  • the compounds in accordance with the invention inhibited HCN4 currents with IC 50 -values between 0.03 ⁇ M and 0.58 ⁇ M.
  • ivabradine revealed an IC 50 of 4.46 ⁇ M, which demonstrates for ivabradine on target level being 7.7- to 149-fold less potent than the compounds in accordance with the invention (i.e. the tested compounds A, B, C, D, E and F).
  • the pyrazole compounds X and Y with certain structural similarities to the tested compounds of the invention were not identified as inhibitors of the HCN4 current.
  • the whole-cell patch-clamp technique was used to investigate the effect of Compound A on the currents of Na v 1.5, K v 1.5, Ca v 1.2 and HCN4 channels, expressed in CHO cells (ATCC, CCL-61).
  • Compound A was tested at concentrations of 200 nM, 600 nM, 2000 nM and 6000 nM.
  • 0.1% DMSO was used as vehicle and ZD7288 as suitable blockers were applied to assure accuracy of the method.
  • the selection antibiotics can be dervied from the Table below: Ion channel Host cell Selection antibiotics Na v 1.5; CHO Geneticin, Puromycin Ca v 1.2 CHO Hygromycin, Geneticin, and Zeocin K v 1.5 CHO Hygromycin HCN4 CHO Hygromycin, Geneticin
  • CHO cells were passaged at a confluence of about 50 - 80 %.
  • cells were seeded onto 35 mm sterile culture dishes containing 2 mL culture complete medium without antibiotics. Cells were cultivated at a density that enables the recording of single cells (without visible connections to other cells) to avoid electrical coupling.
  • Conditions Standard Laboratory Conditions. Cells were incubated at 37°C in a humidified atmosphere with 5 % CO 2 (rel. humidity about 95 %). Culture media: The cells were continuously maintained in and passaged sterile culture flasks containing cell culture medium supplemented with 10% fetal bovine serum and 1.0 % Penicillin/Streptomycin solution. Medium for CHO: F12 (HAM) with L-Glutamine Selection antibiotics: The complete medium as indicated above is supplemented with antibiotics as given in the Table above.
  • Dose levels were as indicated below. Compound A was weighed into a container on a calibrated precision balance and vehicle added to achieve a stock concentration of 6.0 mM (storage at 19°C to 30°C).
  • test solutions were stored at 19°C to 30°C (room temperature). Test solutions were prepared freshly every day. Test solutions were prepared shortly before experimentation. During this period, there were no observations indicating any instability of test items dissolved in bath solution.
  • Concentrations of 200 nM, 600 nM, 2000 nM and 6000 nM of Compound A were tested. They were diluted from the stock solution using bath solution shortly prior to the electrophysiological experiments and kept at room temperature (19°C to 30°C) when in use. The vehicle concentration was adjusted in all dose formulations to 0.1 % DMSO. Frequency of preparation: daily. Storage of dose formulation: On the day of experimentation dose formulations were maintained at room temperature (19 °C - 30 °C) when in use.
  • the 1x bath solution was prepared by diluting 10x bath solution without glucose and 100x glucose solution with water at least every 7 days. Both stock solutions have been prepared prior to the experimental start of the present study and were stored at 1°C to 9°C (10x bath solution) or -10°C to -30°C (100x Glucose solution). When in use, the 1x bath solution was kept at room temperature (19°C - 30°C). When not in use, the 1 x bath solutions were stored at 1°C to 9°C.
  • the final bath solutions included the components: NaCl 137 mM; KCI 4 mM; CaCl 2 1.8 mM; MgCl 2 1 mM; HEPES 10 mM; D-Glucose 10 mM; pH (NaOH) 7.4.
  • the following 1x bath solution was kept at room temperature (19°C - 30°C) when in use. When not in use, the 1 x bath solution was stored at 1°C to 9°C. Bay K8644 was added on the day of experimentation.
  • the final bath solution included the components: NaCl 100 mM; KCI 4.0 mM; NMDG 40 mM; CaCl 2 5.0 mM; MgCl 2 1.0 mM; HEPES 10 mM; D-Glucose 5 mM; Sorbitol 2.50 mM; Bay K8644 0.4 ⁇ M; pH (HCl) 7.4.
  • the 1x pipette solution was thawed every day out of a frozen 1x pipette solution, which had been prepared prior to the experimental start of the present study, aliquoted and stored at -10°C to -30°C. When in use, the 1x pipette solution was filled into the patch-pipettes and kept at room temperature (19-30°C).
  • the 1x pipette solution will include the components outlined below:
  • Ca v 1.2 CsF 135 mM; NaCl 10 mM; CaCl 2 0.2 mM; HEPES 10 mM; EGTA 5 mM; pH (KOH) 7.3.
  • HCN4 KCI 135 mM; MgCl 2 2.5 mM; CaCl 2 2.4 mM; Na 2 ATP 3.0 mM; EGTA 5.0 mM; HEPES 5.0 mM; pH (KOH) 7.3.
  • test compound A was tested to investigate the effects of test Compound A on Na v 1.5, Ca v 1.2, K v 1.5, and HCN4 channel activity.
  • the 35 mm culture dishes upon which cells were seeded at a density allowing single cells to be recorded were placed on the dish holder of the microscope and continuously perfused (at approximately 1 mL/min) with the bath solution described in section 4.7. All solutions applied to cells including the pipette solutions were maintained at room temperature (19°C - 30°C). After formation of a Gigaohm seal between the patch electrodes and individual cells (pipette resistance range: 2.0 M ⁇ - 7.0 M ⁇ ; seal resistance range: > 1 G ⁇ ) the cell membrane across the pipette tip was ruptured to assure electrical access to the cell interior (whole-cell patch-configuration).
  • Ion channel Holding potential Pulse / Duration Frequency Na v 1.5 State dependence -120 / -80mV / 0 mV (resting, fast / slow inactivated state) 0 mV / 10 ms (resting, fast / slow inactivated state) 0.03 Hz (between sweeps at one concentration) 0.0083 Hz (between concentrations) Na v 1.5 use dependence -100 mV -10 mV / 50 ms 1 Hz K v 1.5 -80 mV +40 mV / 5000 ms 0.05 Hz Ca v 1.2 -80 mV / -50 mV repulse (100 ms) 0 mV / 200 ms 0.2 Hz HCN4 -40 mV -120 mV / 2000 ms 0.05 Hz
  • IC 50 values were calculated, if the current amplitude was reduced by more than 50 % at ⁇ 6 ⁇ M of Compound A. As shown in FIG. 17 , only for HCN4 and Na v 1.5 (fast inactivated state) a reduction of the current amplitude of more than 30 % was observed. For NaV1.5 there was a moderate reduction of the currents determined for the fast inactivated state observed, however, the IC 50 was above the highest of 6,000 nM. For HCN4 an IC 50 of 878.81 nM (Hill coefficient 1.15) was determined.
  • IC 50 -values may vary in dependency of the specific measurement conditions applied, specifically in case of a complex patch-clamp setup.
  • the whole-cell patch-clamp technique was used to demonstrate the effects of Compound A on the currents of Na v 1.5, K v 1.5, Ca v 1.2 and HCN4 channels expressed in CHO cells.
  • 0.1% DMSO was used as vehicle and suitable blockers were applied to assure accuracy of the method.
  • Compound A was tested at concentrations of 200 nM, 600 nM, 2000 nM and 6000 nM.
  • the Compound A stock solutions were prepared freshly every day of experimentation. Application of Compound A to the CHO cells was completed within one day. During this period, there was no observation indicating any instability of Compound A solved in vehicle. The effects observed on Na v 1.5, K v 1.5, Ca v 1.2 and HCN4 channels of the Compound A dose formulations analyzed remained stable over several hours at room temperature. During this period, there were no observations indicating any instability of tested Compound A solved in bath solution.
  • the compound A represents the free form of the active drug substance.
  • the dose levels indicated below are expressed in terms of the free base.
  • a correction factor of 1.087 was used when calculating quantities of the tested compound required for dose preparation.
  • Main study animals 24 animals (12 males and 12 females); 3 animals/sex/group.
  • Recovery animals 8 animals (4 males and 4 females); 2 animals/sex for groups 1 and 4.
  • the food was prepared by mixing the weighed pellets with a constant volume of water.
  • the dogs were kept singly or by twos in kennels maintained at a temperature, which ranged between 17°C and 25 °C. Each kennel had an inside yard and outside yard with a total floor space of 9 m 2 . The cages were specially equipped to keep the animals singly during the daily examinations. The outside yard was accessible at all times (except at the time of feeding and examinations).
  • test compound A-vehicle formulations were freshly prepared before each administration.
  • the vehicle (pure Labrafil M1944CS) was stirred well to ensure homogeneity before use.
  • the suspensions were kept under agitation until administration to the dogs.
  • test compound A-formulation of the appropriate concentration was administered by oral administration at a constant administration volume/kg b.w.
  • the first administration of the day was carried out approximately 2 hours before start of feeding. While still in position, the stomach tube was rinsed with 10 mL vehicle immediately after the administration. The control animals received the vehicle at the same administration volume twice daily in the same way as the animals of group 4.
  • the dose levels for the 4-week dog study were set at 3, 10, and 2 x 50 mg/kg.
  • the dose levels for this study have been selected based on the results of the 10-day dose-range-finding study in dogs (LPT Study No. 31586) and available (kinetic) data from single dose studies.
  • the dose increase from 30 to 100 mg/kg did not reveal a further increase in exposure. Dosing 100 mg/kg over 2 separated 50 mg/kg doses 8 hours apart, did lead to a small increase in exposure.
  • the dose levels for the 4-week dog study are set at 3 (human equivalent dose), 10 and 2 x 50 mg/kg.
  • the animals were allocated to the 4 test groups employing a pseudo-random body weight stratification procedure that yields groups with approximately equal mean body weight.
  • Group Compound A dose [mg/kg b.w.] # Dose volume [mL/kg b.w.] Frequency of administration Number and sex of animals MS + RP Animal number MS RP 1 0 2 x 1.25 Twice daily in an 8-h interval 3 + 2 m 1 - 3 4, 5 Control (Vehicle) 3 + 2 f 6 - 8 9, 10 2 3 2.5 Once daily 3 m 11 - 13 none Low dose 3 f 14 - 16 3 10 2.5 Once daily 3 m 17 - 19 none Mid dose 3 f 20 - 22 4 2x50 2 x 1.25 Twice daily in an 8-h interval 3 + 2 m 23 - 25 26, 27 High dose, b.i.d.
  • test compound A represents the free form of the active drug substance. The dose levels are expressed in terms of the free base. Dosing was based on a nominal compound A purity of 100%. A correction factor of 1.087 was applied.
  • the animals were observed individually before and after dosing at each time of dosing for any signs of behavioural changes, reaction to treatment or illness.
  • Parameters were monitored telemetrically with the EMKA Technologies System continuously starting approximately 24 hours prior to the first treatment (baseline; pre-dose values) and for approximately 24 hours in test week 4 and at the end of the recovery period in test week 8.
  • the dogs were habituated and trained for wearing the jackets equipped with the telemetry transmitter and air-filled cuff placed on their tail.
  • ECG, skin temperature, respiratory and physical activity data were recorded continuously. Blood pressure data were transmitted in four consecutive 2-minute intervals followed by a 12-minute pause.
  • the report includes the data recorded at the following intervals:
  • the ECG was evaluated at a time sufficiently before or after the scheduled blood sampling time as blood sampling can cause a disturbance in the ECG recordings.
  • Heart rate (beats/min) PQ-interval (ms) (equivalent to PR interval) QRS-interval (ms) QT-interval (ms) RR-interval (ms)
  • the non-invasive telemetry evaluation was only performed predose and after the 4-week treatment period, and at the end of the recovery period (week 8). Effects are described based on a comparison with the placebo treated animals.
  • Non-invasive telemetry (only week 4 and week 8)
  • the heart rate, the RR-intervals, and the QT-intervals of the previously high dosed dogs were again within the range of the control group values (see Figures 18 to 23 ).
  • Heart rates, RR intervals, QT, and QTc intervals are shown graphically in the following Figures: Figure 18 (Heart rate - males), Figure 19 (Heart rate - females), Figure 20 (RR interval - males), Figure 21 (RR interval - females), Figure 22 (QT interval - males), Figure 23 (QT interval - females).
  • QTc intervals are depicted in the Figures 24 to 26 .
  • beagle dog experiments demonstrate the specific cardiovascular effect of compound A, namely the sole reduction of heart rate, when administered daily by oral administration to beagle dogs for 4 weeks. A complete reversibility of these changes was noted at the end of the 4-week treatment-free recovery period.
  • Cardiovascular parameters are monitored by telemetry in a suitable in vivo model. Control and three escalating dose levels for each animal were tested. Each animal received all doses (control and escalating doses (low, intermediate, high dose) separated by a washout phase (each animal serves as its own control).
  • Formac's silica-based drug delivery technology for the solubility-enhancing formulation of the otherwiese poorly water-soluble compound A as active pharmaceutical ingredient (API) has been tested.
  • Solubility determination First solubility of compound A was determined in different solvents in order to impregnate the drug onto the silica material.
  • Impregnation Compound A was loaded onto OMS-7 via solvent-based impregnation. Briefly, this process involves the addition of a solution of the compound A to the silica material, followed by homogenisation of the wet powder mass and evaporation of the solvent by vacuum drying (50 mbar, 40 °C, 20 hours).
  • Stability studies The silica particles of compound A obtained at three loadings (20 %, 30 % and 40 %) were subjected to stability studies (at 40 °C/75 % RH) for 1 month.
  • Loadability of compound A on OMS-7 Three solvent systems were evaluated in terms of impregnation; i.e. 100 % formic acid, 50/50 % formic acid/methylene chloride and 50/50 % formic acid/acetone. Compound A was impregnated onto OMS-7 to obtain API loadings of 20 %, 30 % and 40 %, after which the samples were assessed for amorphicity by DSC.
  • In vitro dissolution The in vitro dissolution profiles of crystalline compound A and the compound A-OMS-7 powders are depicted in FIG. 31 .
  • An exemplary suspension formulation with Capmul ® and Labrafil in accordance with the invention comprises:
  • Example 7 Pharmacokinetic parameters of compound A administered in various formulations to mice and dogs
  • Compound A was orally administered as non-micronized material, micronized material and ethane sulfonate of compound A suspended in Labrafil ® M1944CS and as non-micronized material in three further formulations to mice and dogs in order to demonstrate the resultant systemic exposure; i.e. pharmacokinetic data.
  • compound A was formulated as suspension in Labrafil ® M1944CS, mesoporous silica particles, aqueous nanosuspension and co-solvent based solution.
  • Single doses of 30 or 500 mg/kg were exemplarily administered to mice, except for the co-solvent based suspension, which was administered at 100 or 500 mg/kg, respectively.
  • a single oral dose of 30 or 100 mg/kg was applied to dogs. Pharmacokinetic parameters were assessed and compared.
  • Compound A was suspended in Labrafil ® M1944CS at 3 and 50 mg/mL for the application of 30 and 500 mg/kg to mice. Compound A was suspended in Labrafil ® M1944CS at 6 and 50 mg/mL for the application of 30 or 100 mg/kg to dogs.
  • Micronized compound A was suspended in Labrafil ® M1944CS at 6 mg/mL for the application of 30 mg/kg to dogs.
  • Compound A ethane sulfonate was suspended in Labrafil ® M1944CS at 15 mg/mL for the application of 30 mg/kg to dogs.
  • Silica powder was weighed and suspended in a vehicle solution composed of 0.5% Poloxamer 188 and 0.5% sodium lauryl sulfate. The suspension was gently stirred (magnetic stir bar) for approx. 2 min until all powder was wetted and a homogeneous, brown, opaque suspension was obtained. Visible lumps were destroyed by sonication. The suspension was gently stirred until administration to avoid sedimentation.
  • a suspension with a final concentration of 10.7 mg silica powder/mL suspension and 3 mg compound A/mL were prepared to apply 30 mg/kg to mice.
  • Suspensions with final concentration of 179.2 mg silica powder/mL suspension and 50 mg compound A/mL were prepared to apply 500 mg/kg to mice.
  • Suspensions with final concentration of 17.7 mg silica powder/mL suspension and 20 mg compound A/mL were prepared to apply 100 mg/kg to dogs.
  • the supplied sterile nanosuspension contained 81.76 mg compound A/g in a vehicle solution composed of 0.034% Poloxamer 188 and 0.5% PVP K17.
  • the suspension was vortex-mixed for 5 min and subsequently diluted in sterile vehicle to obtain a nanosuspension of 3 mg compound A/g for the low dose group (30 mg/kg) and 50 mg compound A/g for the high dose group (500 mg/kg) in mice.
  • the suspension was vortex-mixed for 5 min and subsequently diluted in sterile vehicle to obtain a nanosuspension of 30 mg compound A/g for the administration of 100 mg/kg to dogs.
  • the nanosuspension was subjected to sonication for 30 min with intermittent shaking to avoid particle agglomeration.
  • the syringe was rinsed with the nanosuspension 2-3 times prior to injection. The exact amount of administered nanosuspension was determined by weighting the syringe before and after dosage.
  • a ready-to-use vehicle consisting of acetic acid and Capmul MCM C8 EP in a ratio of 14.3 % + 85.7 % (w+w) was supplied.
  • the pH of the final formulation was around 2.5.
  • Blood sampling time points were kept constant for all formulations at 1, 2, 4, 8, 24, 48, 72 and 120 hours after administration of 30 mg/kg or 100 mg/kg to mice.
  • mice were sacrificed per time point.
  • Blood sampling time points of the dogs were predose 0.5, 1, 2, 4, 8, 12, 24, 48, 72, 96, 120, 144, 168, 192, 216, 264 and 288 hours after administration of a Labrafil ® M1944CS suspension and predose 0.5, 1, 2, 4, 8, 12, 24, 48, 72, 96, 144, 192, 240 and 288 hours after administration of all other formulations.
  • Plasma concentrations were obtained by LC-MS/MS measurements.
  • Pharmacokinetic parameters were calculated by use of the PhoenixTM WinNonLin ® software (Build 6.3.0.395, Certara L.P.) using non-compartmental pharmacokinetic models 200-202 (extravascular input) and linear trapezoidal fit (linear Interpolation). All further calculations were carried out with Microsoft Excel 2010.
  • the objective was to demonstrate the systemic exposure of compound A after administration in the pharmaceutical formulations of the invention by comparison of compound A formulated as Labrafil ® M1944CS suspension, mesoporous silica particles, aqueous nanosuspension or co-solvent based suspension after administration to female CD-1 mice.
  • the Labrafil ® M1944CS suspension showed slightly higher exposure with 234 h*mg/L when compared to mesoporous silica particles with 181 h*mg/L. Maximal plasma concentrations (C max ) were observed 8 hours after administration of the Labrafil ® M1944CS suspension at 4.22 mg/kg and 24 hours after administration of the mesoporous silica particles at 4.47 mg/L.
  • the aqueous nanosuspension showed clearly lower values for AUC0- ⁇ and Cmax with 137 h*mg/kg and 3.62 mg/L.
  • the aqueous nanosuspension was very foamy and the correct dose was determined by weighing the application syringe before and after administration. The applied doses were found to be within a range of 25.3 to 31.7 mg/kg and were in good agreement with the nominal dose of 30 mg/kg
  • the mesoporous silica particles showed slightly higher exposure (AUC 0- ⁇ ) and plasma concentrations when compared to the Labrafil ® M1944CS suspension but also higher inter-individual variability. Exposure was found to be 567 and 644 h*mg/kg for the Labrafil ® M1944CS suspension and the mesoporous silica particles, respectively. C max accounted for 7.43 mg/L and 7.57 mg/L for compound A administered as Labrafil ® M1944CS suspension and mesoporous silica particles.
  • AUC0- ⁇ was 346 h*mg/L and Cmax was 7.56 mg/L.
  • the objective was to demonstrate the highest exposure by comparison of the pharmaceutical formulations of the inventions with compound A; being it micronized compound A, compound A mono ethane sulfonate and non-micronized compound A after administration at 30 mg compound A/kg to dogs.
  • non-micronized compound A formulated as Labrafil ® M1944CS suspension, mesoporous silica particles, or aqueous nanosuspension. All formulations were administered at 100 mg/kg to dogs.
  • the corresponding exposure in terms of AUC 0- ⁇ was 190, 218 and 297 h*mg/L for micronized compound A, ethane sulfonate and non-micronized compound A.
  • two treatments micronized compound A and compound A mono ethane sulfonate
  • vomiting occurred in two to three out of four animals which does not exclude, that the exposure would have been even higher if no vomiting had occurred.
  • Comparable plasma concentration vs. time profiles were obtained after oral administration of 100 mg compound A/kg body weight to dogs formulated as either Labrafil ® M1944CS suspension or mesoporous silica particles. Accordingly, similar exposure was obtained. Maximal plasma concentrations were observed 24 to 48 hours after administration of 100 mg compound A/kg as Labrafil ® M1944CS suspension at 2.36 mg/mL and a corresponding exposure in terms of AUC 0- ⁇ of 207 h*mg/L. With compound A formulated as mesoporous silica particles maximal plasma concentrations were found 8 to 24 hours after administration at 1.86 mg/L and an AUC 0- ⁇ of 208 h*mg/L.
  • Charts on mean plasma concentration versus time profiles of the above-described examples can be derived from the Figures 38 to 45 .
  • excipient(s) or similar terms herein means any substance, not itself a therapeutic agent, used as a diluent, adjuvant, or vehicle for delivery of a therapeutic agent to a subject or added to a pharmaceutical composition to improve its handling or storage properties or to permit or facilitate formation of a solid dosage form such as a tablet, capsule, or a solution or suspension suitable for oral, parenteral, intradermal, subcutaneous, or topical application.
  • Excipients can include, by way of illustration and not limitation, solubizers, diluents, disintegrants, binding agents, adhesives, wetting agents, polymers, lubricants, glidants, stabilizers, and substances added to mask or counteract a disagreeable taste or odor, flavors, dyes, fragrances, and substances added to improve appearance of the composition.
  • Acceptable excipients include (but are not limited to) stearic acid, citric acid, maleic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, magnesium carbonate, talc, gelatin, acacia gum, sodium alginate, pectin, dextrin, mannitol, sorbitol, lactose, sucrose, starches, gelatin, cellulosic materials, such as cellulose esters of alkanoic acids and cellulose alkyl esters, low melting wax, cocoa butter or powder, polymers such as polyvinyl-pyrrolidone, polyvinyl alcohol, and polyethylene glycols, inert carriers such as silica particles and other pharmaceutically acceptable materials.
  • the choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • treatment is understood here as synonymous with the term “treatment”.
  • prevention is used in the present invention interchangeably and refer to avoiding or reducing the risk of a disease, ailment, injury or health disorder, an unfolding or progression of such conditions and / or the symptoms of such conditions, to suffer or to have.
  • the treatment or prevention of a disease, condition, an illness, an injury or a health disorder can be partial or complete.
  • references herein to "treat”, “treating”, “treatment” and the like include curative, palliative and prophylactic treatment.
  • pharmaceutically acceptable is meant the carrier, vehicle, or diluent and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • pharmaceutically effective amount refers to an amount of the compound of Formula I (or a combination agent or a Formula I compound in combination with a combination agent) sufficient to treat, prevent onset of or delay or diminish the symptoms and physiological manifestations of the indications described herein.
  • coronary artery disease is selected, but not limited to, the group consisting of atherosclerotic plaque (e.g., prevention, regression, stablilization), vulnerable plaque (e.g., prevention, regression, stabilization), vulnerable plaque area (reduction), arterial calcification (e.g., calcific aortic stenosis), increased coronary artery calcium score, dysfunctional vascular reactivity, vasodilation disorders, coronary artery spasm, first myocardial infarction, myocardia re-infarction, ischemic cardiomyopathy, stent restenosis, PTCA restenosis, arterial restenosis, coronary bypass graft restenosis, vascular bypass restenosis, decreased exercise treadmill time, angina pectoris/chest pain, unstable angina pectoris, exertional dyspnea, decreased exercise capacity, ischemia (reduce time to), silent ischemia (reduce time to), increased severity and frequency of ischemic symptoms, reper
  • hypertension is selected, but not limited to, the group consisting of lipid disorders with hypertension, systolic hypertension and diastolic hypertension.
  • diabetes refers to any of a number of diabetogenic states including type I diabetes, type II diabetes, Syndrome X, Metabolic syndrome, lipid disorders associated with insulin resistance, impaired glucose tolerance, non-insulin dependent diabetes, microvascular diabetic complications, reduced nerve conduction velocity, reduced or loss of vision, diabetic retinopathy, increased risk of amputation, decreased kidney function, kidney failure, insulin resistance syndrome, pluri-metabolic syndrome, central adiposity (visceral)(upper body), diabetic dyslipidemia, decreased insulin sensitization, diabetic retinopathy/neuropathy, diabetic nephropathy/micro and macro angiopathy and micro/macro albuminuria, diabetic cardiomyopathy, diabetic gastroparesis, obesity, increased hemoglobin glycoslation (including HbA1C), improved glucose control, impaired renal function (dialysis, endstage) and hepatic function (mild, moderate, severe).
  • HbA1C hemoglobin glycoslation
  • improved glucose control impaired renal function (dialysis,
  • Methodabolic syndrome also known as “Syndrome X” refers to a common clinical disorder that is defined as the presence of increased insulin concentrations in association with other disorders including viceral obesity, hyperlipidemia, dyslipidemia, hyperglycemia, hypertension, and potentially hyperuricemis and renal dysfunction.
  • heart failure both acute and chronic manifestations of heart failure, as well as specific or related disease forms such as acute congestive heart failure, right heart failure, left ventricular failure, global failure, ischemic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, idiopathic cardiomyopathy, congenital heart disease, heart failure in valvular heart, mitral valve stenosis, mitral regurgitation, aortic stenosis, aortic regurgitation, tricuspid stenosis, tricuspid insufficiency, pulmonary valve stenosis, pulmonary valve insufficiency, combined valvular, heart muscle inflammation (myocarditis), chronic myocarditis, acute myocarditis, viral myocarditis, diabetic heart failure, alcoholic cardiomyopathy, cardiac storage diseases, diastolic congestive heart failure and systolic heart failure and acute phases of exacerbation of chronic heart failure (w
  • slow rate heating or similar expressions denote within the context of the present invention that a chemical reaction or an apparatus and/or device, wherein a chemical reaction takes place, is gradually heated by adequate heating steps to allow for a dosed thermal energy supply depending on the chemical reaction or apparatus and/or device used. For instance, and only exemplarily without being limited thereto, a slow heating rate could mean to increase the temperature of a chemical reaction or an apparatus and/or device by 10 +/- 5 °C per hour.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP16177663.8A 2016-07-01 2016-07-01 Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités Ceased EP3263567A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP16177663.8A EP3263567A1 (fr) 2016-07-01 2016-07-01 Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP16177663.8A EP3263567A1 (fr) 2016-07-01 2016-07-01 Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités

Publications (1)

Publication Number Publication Date
EP3263567A1 true EP3263567A1 (fr) 2018-01-03

Family

ID=56345041

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16177663.8A Ceased EP3263567A1 (fr) 2016-07-01 2016-07-01 Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités

Country Status (1)

Country Link
EP (1) EP3263567A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136804A (en) * 1998-03-13 2000-10-24 Merck & Co., Inc. Combination therapy for treating, preventing, or reducing the risks associated with acute coronary ischemic syndrome and related conditions
WO2005086902A2 (fr) * 2004-03-08 2005-09-22 Wyeth Modulateurs de canal ionique
WO2008042388A1 (fr) * 2006-10-03 2008-04-10 Arena Pharmaceuticals, Inc. Dérivés de pyrazole en tant que modulateurs du récepteur 5ht2a de la sérotonine utiles dans le traitement de troubles liés à ce récepteur
WO2014027112A1 (fr) * 2012-08-17 2014-02-20 Aicuris Gmbh & Co. Kg Tri(hétéro)arylpyrazoles et leur utilisation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136804A (en) * 1998-03-13 2000-10-24 Merck & Co., Inc. Combination therapy for treating, preventing, or reducing the risks associated with acute coronary ischemic syndrome and related conditions
WO2005086902A2 (fr) * 2004-03-08 2005-09-22 Wyeth Modulateurs de canal ionique
WO2008042388A1 (fr) * 2006-10-03 2008-04-10 Arena Pharmaceuticals, Inc. Dérivés de pyrazole en tant que modulateurs du récepteur 5ht2a de la sérotonine utiles dans le traitement de troubles liés à ce récepteur
WO2014027112A1 (fr) * 2012-08-17 2014-02-20 Aicuris Gmbh & Co. Kg Tri(hétéro)arylpyrazoles et leur utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KONCZ I ET AL: "Selective pharmacological inhibition of the pacemaker channel isoforms (HCN1-4) as new possible therapeutical targets", CURRENT MEDICINAL CHEMISTRY-CARDIOVASCULAR & HEMATOLOGICAL AGENTS,, vol. 18, no. 24, 1 August 2011 (2011-08-01), pages 3662 - 3674, XP008160113, ISSN: 0929-8673 *

Similar Documents

Publication Publication Date Title
TWI516263B (zh) 經取代之5-氟-1h-吡唑并吡啶類及其用途
ES2547153T3 (es) Derivados de 8-alcoxi-aminotetralina sustituidos y su uso
JP6324956B2 (ja) 置換アミノインダン−およびアミノテトラリンカルボン酸ならびにその使用
EP1988091B1 (fr) Composés tricycliques
EP3004094B1 (fr) Imidazo[1,2-a]pyridines à substitution 3-aryle et leur utilisation
JP5897566B2 (ja) 環式n,n’−ジアリールチオ尿素及びn,n’−ジアリール尿素−アンドロゲン受容体アンタゴニスト、抗癌剤、その調製のための方法及び使用
JP2021127346A (ja) ジアザ二環式置換イミダゾピリミジンおよび呼吸障害を治療するためのその使用
EP3194386A2 (fr) Stimulateurs de sgc
TW200938200A (en) Methyl-substituted piperidine derivative
EP2878594B1 (fr) Composé de difluorométhylène
KR20080036041A (ko) 아세틸 조효소 카복실라제(acc) 억제제로서의스피로크로마논 유도체
JP2017538676A (ja) ミトコンドリア透過性遷移孔(mtPTP)の小分子阻害剤
CA2943005A1 (fr) Imidazo[1,2-a]pyridine-carboxamides cyano-substitues et leur utilisation
JP2016527295A (ja) 置換イミダゾ[1,2−a]ピラジンカルボキサミドおよびその使用
EP4171566A1 (fr) Composés de liaison au céréblon, compositions de ceux-ci et procédés de traitement avec ceux-ci
WO2022105852A1 (fr) Dérivé de triazine dione, son procédé de préparation et son application en médecine
AU2015288381A1 (en) Substituted 2-thioxo-imidazolidin-4-ones and spiro analogues thereof, active anti-cancer ingredient, pharmaceutical composition, medicinal preparation, method for treating prostate cancer
BRPI0618284A2 (pt) derivado de pirazolo-[4,3-d]-pirimidin-5-il usado como inibidores de pde5
JP2013515766A (ja) イマチニブジクロロ酢酸塩及びそれを含む抗癌剤組成物
JP6908805B2 (ja) Cnsおよび他の障害の処置のためのベンゾフラン誘導体
US9428460B2 (en) N-[4-(quinolin-4-yloxy)cyclohexyl(methyl)](hetero)arylcarboxamides as androgen receptor antagonists, production and use thereof as medicinal products
JP2017514901A (ja) 6−クロロ置換イミダゾ[1,2−a]ピリジンカルボキサミドおよびその使用
EP3263567A1 (fr) Pyrazoles substitués avec une carboxamide et tri(hétéro)aryle-pyrazoles pour utilisation dans le traitement et/ou prévention des maladies cariovasculaires et/ou leurs comorbidités
EP3227287B1 (fr) Imidazo[1,2-a]pyridines à substitution hétéroaryle et leur utilisation
TW201245180A (en) Novel phenylpyridine derivatives and medicament comprising the derivatives

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20180210